195 results on '"Dewerchin M"'
Search Results
2. Vasculaire endotheliale groeifactor en hypoxie-induceerbare factor 2α: drijvende krachten achter longmaturatie; belang voor respiratoir distress syndroom
- Author
-
Compernolle, V., Dewerchin, M., Moons, L., and Carmeliet, P.
- Published
- 2005
- Full Text
- View/download PDF
3. Variable region heavy chain glycosylation determines the anticoagulant activity of a factor VIII antibody
- Author
-
JACQUEMIN, M., RADCLIFFE, C. M., LAVENDʼHOMME, R., WORMALD, M. R., VANDERELST, L., WALLAYS, G., DEWAELE, J., COLLEN, D., VERMYLEN, J., DWEK, R. A., SAINT-REMY, J-M, RUDD, P. M., and DEWERCHIN, M.
- Published
- 2006
4. BLOOD PRESSURE IN MICE WITH A TARGETED DELETION OF THE SOLUBLE GUANYLYL CYCLASE ALPHA1 SUBUNIT: S1O9
- Author
-
Brouckaert, P., Buys, E., Sips, P., Rogge, E., Dewerchin, M., and Cauwels, A.
- Published
- 2005
5. Inhibition of factor VIII with a partially inhibitory human recombinant monoclonal antibody prevents thrombotic events in a transgenic model of type II HBS antithrombin deficiency in mice
- Author
-
Dewerchin, M., Van Der Elst, L., Singh, I., Grailly, S., Saint‐Remy, J‐M., Collen, D., and Jacquemin, M.
- Published
- 2004
- Full Text
- View/download PDF
6. Endogenous α2-antiplasmin does not enhance glomerular fibrin deposition or injury in glomerulonephritis
- Author
-
Kitching, A.R., Turner, A.L., O'Sullivan, K-M., Dewerchin, M., and Lijnen, H.R.
- Published
- 2003
- Full Text
- View/download PDF
7. Vascular and cellular proteolytic activity in mice with α2-antiplasmin gene inactivation
- Author
-
Lijnen, H.R., Ugwu, F., Maquoi, E., Lemmens, G., Hoef, B.Van, Dewerchin, M., and Collen, D.
- Published
- 2000
- Full Text
- View/download PDF
8. α2-Antiplasmin Gene Deficiency in Mice Is Associated With Enhanced Fibrinolytic Potential Without Overt Bleeding
- Author
-
Lijnen, H.R., Okada, K., Matsuo, O., Collen, D., and Dewerchin, M.
- Published
- 1999
- Full Text
- View/download PDF
9. Loss of HIF-2[alpha] and inhibition of VEGF impair fetal lung maturation, whereas treatment with VEGF prevents fatal respiratory distress in premature mice
- Author
-
Compernolle, V., Brusselmans, K., Acker, T., Hoet, P., Tjwa, M., Beck, H., Plaisance, S., Dor, Y., Keshet, E., Lupu, F., Nemery, B., Dewerchin, M., Van Veldhoven, P., Plate, K., Moons, L., Collen, D., and Carmeliet, P.
- Abstract
Author(s): V. Compernolle; K. Brusselmans; T. Acker; P. Hoet; M. Tjwa; H. Beck; S. Plaisance; Y. Dor; E. Keshet; F. Lupu; B. Nemery; M. Dewerchin; P. Van Veldhoven; K. Plate; [...]
- Published
- 2002
- Full Text
- View/download PDF
10. Soluble guanylate cyclase-alpha1 deficiency selectively inhibits the pulmonary vasodilator response to nitric oxide and increases the pulmonary vascular remodeling response to chronic hypoxia.
- Author
-
Vermeersch P, Buys E, Pokreisz P, Marsboom G, Ichinose F, Sips P, Pellens M, Gillijns H, Swinnen M, Graveline A, Collen D, Dewerchin M, Brouckaert P, Bloch KD, Janssens S, Vermeersch, Pieter, Buys, Emmanuel, Pokreisz, Peter, Marsboom, Glenn, and Ichinose, Fumito
- Published
- 2007
11. Insights in Vessel Development and Vascular Disorders Using Targeted Inactivation and Transfer of Vascular Endothelial Growth Factor, the Tissue Factor Receptor, and the Plasminogen System.
- Author
-
CARMELIET, PETER, MOONS, LIEVE, DEWERCHIN, M., MACKMAN, NIGEL, LUTHER, THOMAS, BREIER, GEORG, PLOPLIS, V., MÜLLER, M., NAGY, A., PLOW, E., GERARD, R., EDGINGTON, THOMAS, RISAU, W., and COLLEN, DÉSIRÉ
- Published
- 1997
- Full Text
- View/download PDF
12. Generation of Pex5-loxP mice allowing the conditional elimination of peroxisomes.
- Author
-
Baes, M., Dewerchin, M., Janssen, A., Collen, D., and Carmeliet, P.
- Published
- 2002
- Full Text
- View/download PDF
13. Validation of an enzyme-linked immunosorbent assay for the quantification of human IgG directed against the repeat region of the circumsporozoite protein of the parasite Plasmodium falciparum
- Author
-
Clement Frederic, Dewar Vincent, Van Braeckel Eva, Desombere Isabelle, Dewerchin Marianne, Swysen Christine, Demoitié Marie-Ange, Jongert Erik, Cohen Joe, Leroux-Roels Geert, and Cambron Pierre
- Subjects
Malaria ,Plasmodium falciparum ,Circumsporozoite protein ,Enzyme-linked immunosorbent assay ,R32LR ,Validation ,Arctic medicine. Tropical medicine ,RC955-962 ,Infectious and parasitic diseases ,RC109-216 - Abstract
Abstract Background Several pre-erythrocytic malaria vaccines based on the circumsporozoite protein (CSP) antigen of Plasmodium falciparum are in clinical development. Vaccine immunogenicity is commonly evaluated by the determination of anti-CSP antibody levels using IgG-based assays, but no standard assay is available to allow comparison of the different vaccines. Methods The validation of an anti-CSP repeat region enzyme-linked immunosorbent assay (ELISA) is described. This assay is based on the binding of serum antibodies to R32LR, a recombinant protein composed of the repeat region of P. falciparum CSP. In addition to the original recombinant R32LR, an easy to purify recombinant His-tagged R32LR protein has been constructed to be used as solid phase antigen in the assay. Also, hybridoma cell lines have been generated producing human anti-R32LR monoclonal antibodies to be used as a potential inexhaustible source of anti-CSP repeats standard, instead of a reference serum. Results The anti-CSP repeats ELISA was shown to be robust, specific and linear within the analytical range, and adequately fulfilled all validation criteria as defined in the ICH guidelines. Furthermore, the coefficient of variation for repeatability and intermediate precision did not exceed 23%. Non-interference was demonstrated for R32LR-binding sera, and the assay was shown to be stable over time. Conclusions This ELISA, specific for antibodies directed against the CSP repeat region, can be used as a standard assay for the determination of humoral immunogenicity in the development of any CSP-based P. falciparum malaria vaccine.
- Published
- 2012
- Full Text
- View/download PDF
14. Corrigendum: Loss of HIF-2α and inhibition of VEGF impair fetal lung maturation, whereas treatment with VEGF prevents fatal respiratory distress in premature mice.
- Author
-
Compernolle, V., Brusselmans, K., Acker, T., Hoet, P., Tjwa, M., Beck, H., Plaisance, S., Dor, Y., Keshet, E., Lupu, F., Nemery, B., Dewerchin, M., Van Veldhoven, P., Plate, K., Moons, L., Collen, D., and Carmeliet, P.
- Subjects
LUNG diseases ,PERIODICALS - Abstract
Presents a correction to an error made in the article 'Loss of HIF-2alpha and inhibition of VEGF fetal lung maturation, whereas treatment with VEGF prevents fatal respiratory distress in premature mice,' by V. Compernolle et al published in 'Nature Medicine' journal in 2002.
- Published
- 2002
- Full Text
- View/download PDF
15. Characterisation of a chemical conjugate between a low molecular weight form of recombinant single chain urokinase-type plasminogen activator (comprising Leu 144 through Leu 411) and F(ab′) 2-fragments of a fibrin D-dimer-specific monoclonal antibody
- Author
-
Dewerchin, M., Lijnen, H.R., and Collen, D.
- Published
- 1990
- Full Text
- View/download PDF
16. Characterisation of conjugates of thrombin-treated single chain urokinase-type plasminogen activator with a monoclonal antibody specific for crosslinked fibrin
- Author
-
Dewerchin, M., Lijnen, H.R., Van Hoef, B., De Cock, F., and Collen, D.
- Published
- 1990
- Full Text
- View/download PDF
17. Deficiency of vascular endothelial growth factor-D does not affect murine adipose tissue development
- Author
-
Lijnen, H.R., Frederix, L., Van Hoef, B., and Dewerchin, M.
- Subjects
- *
VASCULAR endothelial growth factors , *ADIPOSE tissues , *OBESITY , *BODY weight , *LABORATORY mice , *NEOVASCULARIZATION , *GENE expression - Abstract
Abstract: Vascular endothelial growth factor (VEGF)-D deficiency had no significant effect on total body weight or on subcutaneous (SC) or gonadal (GON) adipose tissue mass of mice kept on a standard fat (SFD) or a high fat diet (HFD) for 15 weeks. The composition of SC and GON adipose tissues of VEGF-D deficient mice in terms of size and density of adipocytes or blood vessels was also comparable to that of wild-type control mice. Staining of lymphatic vessels in adipose tissue sections did not reveal marked differences between both genotypes. The absence of an effect of VEGF-D deficiency could not be explained by compensatory increases of VEGF-C expression in adipose tissues of the deficient mice. Thus, our data do not support an important role of VEGF-D in (lymph) angiogenesis or in adipose tissue development. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
18. 190 Characterization and targeting of the murine α2-antiplasmin gene
- Author
-
Lijnen, H.R., Dewerchin, M., Okada, K., Belayew, A., and Collen, D.
- Published
- 1997
- Full Text
- View/download PDF
19. 78 Receptor-independent role of urokinase-type plasminogen activator during vascular wound healing in mice
- Author
-
Carmeliet, P., Moons, L., Dewerchin, M., Rosenberg, S., Herbert, J.-M., Lupu, F., and Collen, D.
- Published
- 1997
- Full Text
- View/download PDF
20. 33: Adenovirus-Mediated Overexpression of Human t-PA Prevents Peritoneal Adhesion Formation/Reformation in Rats
- Author
-
Atta, H.M., Al-Hendy, A., El-Rehany, M.A., Abdel Raheim, S.R., Abd Elghany, H.M., Foad, R., and Dewerchin, M.
- Published
- 2009
- Full Text
- View/download PDF
21. Single-cell RNA sequencing of cystic fibrosis liver disease explants reveals endothelial complement activation.
- Author
-
Declercq M, Treps L, Geldhof V, Conchinha NV, de Rooij LPMH, Subramanian A, Feyeux M, Cotinat M, Boeckx B, Vinckier S, Dupont L, Vermeulen F, Boon M, Proesmans M, Libbrecht L, Pirenne J, Monbaliu D, Jochmans I, Dewerchin M, Eelen G, Roskams T, Verleden S, Lambrechts D, Carmeliet P, and Witters P
- Subjects
- Humans, Liver pathology, Liver metabolism, Male, Female, Adult, Liver Cirrhosis genetics, Liver Cirrhosis pathology, Liver Diseases genetics, Cystic Fibrosis genetics, Single-Cell Analysis, Endothelial Cells metabolism, Sequence Analysis, RNA, Complement Activation
- Abstract
Background & Aims: Cystic fibrosis (CF) is considered a multisystemic disorder in which CF-associated liver disease (CFLD) is the third most common cause of mortality. Currently, no effective treatment is available for CFLD because its pathophysiology is still unclear. Interestingly, CFLD exhibits identical vascular characteristics as non-cirrhotic portal hypertension, recently classified as porto-sinusoidal vascular disorders (PSVD)., Methods: Since endothelial cells (ECs) are an important component in PSVD, we performed single-cell RNA sequencing (scRNA-seq) on four explant livers from CFLD patients to identify differential endothelial characteristics which could contribute to the disease. We comprehensively characterized the endothelial compartment and compared it with publicly available scRNA-seq datasets from cirrhotic and healthy livers. Key gene signatures were validated ex vivo on patient tissues., Results: We found that ECs from CF liver explants are more closely related to healthy than cirrhotic patients. In CF patients we also discovered a distinct population of liver sinusoidal ECs-coined CF LSECs-upregulating genes involved in the complement cascade and coagulation. Finally, our immunostainings further validated the predominant periportal location of CF LSECs., Conclusions: Our work showed novel aspects of human liver ECs at the single-cell level thereby supporting endothelial involvement in CFLD, and reinforcing the hypothesis that ECs could be a driver of PSVD. Therefore, considering the vascular compartment in CF and CFLD may help developing new therapeutic approaches for these diseases., (© 2024 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
22. The gluconeogenesis enzyme PCK2 has a non-enzymatic role in proteostasis in endothelial cells.
- Author
-
de Zeeuw P, Treps L, García-Caballero M, Harjes U, Kalucka J, De Legher C, Brepoels K, Peeters K, Vinckier S, Souffreau J, Bouché A, Taverna F, Dehairs J, Talebi A, Ghesquière B, Swinnen J, Schoonjans L, Eelen G, Dewerchin M, and Carmeliet P
- Subjects
- Humans, Human Umbilical Vein Endothelial Cells metabolism, Glucose metabolism, Autophagy, Unfolded Protein Response, Phosphoenolpyruvate Carboxykinase (ATP), Proteostasis, Gluconeogenesis genetics, Endothelial Cells metabolism, Phosphoenolpyruvate Carboxykinase (GTP) metabolism, Phosphoenolpyruvate Carboxykinase (GTP) genetics
- Abstract
Endothelial cells (ECs) are highly glycolytic, but whether they generate glycolytic intermediates via gluconeogenesis (GNG) in glucose-deprived conditions remains unknown. Here, we report that glucose-deprived ECs upregulate the GNG enzyme PCK2 and rely on a PCK2-dependent truncated GNG, whereby lactate and glutamine are used for the synthesis of lower glycolytic intermediates that enter the serine and glycerophospholipid biosynthesis pathways, which can play key roles in redox homeostasis and phospholipid synthesis, respectively. Unexpectedly, however, even in normal glucose conditions, and independent of its enzymatic activity, PCK2 silencing perturbs proteostasis, beyond its traditional GNG role. Indeed, PCK2-silenced ECs have an impaired unfolded protein response, leading to accumulation of misfolded proteins, which due to defective proteasomes and impaired autophagy, results in the accumulation of protein aggregates in lysosomes and EC demise. Ultimately, loss of PCK2 in ECs impaired vessel sprouting. This study identifies a role for PCK2 in proteostasis beyond GNG., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
23. COVID-19 plasma induces subcellular remodelling within the pulmonary microvascular endothelium.
- Author
-
Passi R, Cholewa-Waclaw J, Wereski R, Bennett M, Veizades S, Berkeley B, Caporali A, Li Z, Rodor J, Dewerchin M, Mills NL, Beqqali A, Brittan M, and Baker AH
- Subjects
- Humans, SARS-CoV-2, Lung, Endothelium, Endothelial Cells metabolism, COVID-19
- Abstract
Background: COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can affect multiple organ systems, including the pulmonary vasculature. Endothelial cells (ECs) are thought to play a key role in the propagation of COVID-19, however, our understanding of the exact scale of dysregulation sustained by the pulmonary microvasculature (pMV) remains incomplete. Here we aim to identify transcriptional, phenotypic, and functional changes within the pMV induced by COVID-19., Methods and Results: Human pulmonary microvascular endothelial cells (HPMVEC) treated with plasma acquired from patients hospitalised with severe COVID-19 were compared to HPMVEC treated with plasma from patients hospitalised without COVID-19 but with other severe illnesses. Exposure to COVID-19 plasma caused a significant functional decline in HPMVECs as seen by a decrease in both cell viability via the WST-1 cell-proliferation assay and cell-to-cell barrier function as measured by electric cell-substrate impedance sensing. High-content imaging using a Cell Painting image-based assay further quantified morphological variations within sub-cellular organelles to show phenotypic changes in the whole endothelial cell, nucleus, mitochondria, plasma membrane and nucleolus morphology. RNA-sequencing of HPMVECs treated with COVID-19 plasma suggests the observed phenotype may, in part, be regulated by genes such as SMAD7, BCOR, SFMBT1, IFIT5 and ZNF566 which are involved in transcriptional regulation, protein monoubiquitination and TGF-β signalling., Conclusion and Impact: During COVID-19, the pMV undergoes significant remodelling, which is evident based on the functional, phenotypic, and transcriptional changes seen following exposure to COVID-19 plasma. The observed morphological variation may be responsible for downstream complications, such as a decline in overall cellular function and cell-to-cell barrier integrity. Moreover, genes identified through bulk RNA sequencing may contribute to our understanding of the observed phenotype and assist in developing strategies that can inform the rescue of the dysregulated endothelium., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
24. Detailed protocol for a corneal thermal cauterization-based (lymph-)angiogenesis assay in mice.
- Author
-
Truong AK, Becker LM, Dekoning N, Bouché A, Veys K, Hosseinkhani B, Dewerchin M, Eelen G, and Carmeliet P
- Abstract
Angiogenesis and lymphangiogenesis, the formation of new blood or lymphatic vessels, respectively, from preexisting vasculature is essential during embryonic development, but also occurs during tissue repair and in pathological conditions (cancer; ocular disease; ischemic, infectious and inflammatory disorders), which are all characterized to a certain extent by inflammatory conditions. Hence, a rapid, inexpensive, feasible / technically easy, reliable assay of inflammation-induced (lymph-)angiogenesis is highly valuable. In this context, the corneal thermal cauterization assay in mice is a simple, low-cost, reproducible, insightful and labor-saving assay to gauge the role of inflammation in angiogenesis and lymphangiogenesis. However, to the best of our knowledge, there is no standardized protocol to perform this assay. Here, we provide a step-by-step description of the model's procedures, which include:•The thermal cauterization of the corneas,•Enucleation and dissection of the corneas,•Subsequent immunofluorescence staining of the neovasculature, and morphometric analysis. We also discuss ethical considerations and aspects related to animal welfare guidelines. Altogether, this paper will help to increase the reproducibility of the corneal thermal cauterization model and facilitate its use for angiogenesis and lymphangiogenesis research., Competing Interests: The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (© 2023 The Authors. Published by Elsevier B.V.)
- Published
- 2023
- Full Text
- View/download PDF
25. Intracellular BAPTA directly inhibits PFKFB3, thereby impeding mTORC1-driven Mcl-1 translation and killing MCL-1-addicted cancer cells.
- Author
-
Sneyers F, Kerkhofs M, Speelman-Rooms F, Welkenhuyzen K, La Rovere R, Shemy A, Voet A, Eelen G, Dewerchin M, Tait SWG, Ghesquière B, Bootman MD, and Bultynck G
- Subjects
- Myeloid Cell Leukemia Sequence 1 Protein genetics, Egtazic Acid, Phosphofructokinase-2 genetics, Phosphoric Monoester Hydrolases, Neoplasms
- Abstract
Intracellular Ca
2+ signals control several physiological and pathophysiological processes. The main tool to chelate intracellular Ca2+ is intracellular BAPTA (BAPTAi ), usually introduced into cells as a membrane-permeant acetoxymethyl ester (BAPTA-AM). Previously, we demonstrated that BAPTAi enhanced apoptosis induced by venetoclax, a BCL-2 antagonist, in diffuse large B-cell lymphoma (DLBCL). This finding implied a novel interplay between intracellular Ca2+ signaling and anti-apoptotic BCL-2 function. Hence, we set out to identify the underlying mechanisms by which BAPTAi enhances cell death in B-cell cancers. In this study, we discovered that BAPTAi alone induced apoptosis in hematological cancer cell lines that were highly sensitive to S63845, an MCL-1 antagonist. BAPTAi provoked a rapid decline in MCL-1-protein levels by inhibiting mTORC1-driven Mcl-1 translation. These events were not a consequence of cell death, as BAX/BAK-deficient cancer cells exhibited similar downregulation of mTORC1 activity and MCL-1-protein levels. Next, we investigated how BAPTAi diminished mTORC1 activity and identified its ability to impair glycolysis by directly inhibiting 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) activity, a previously unknown effect of BAPTAi . Notably, these effects were also induced by a BAPTAi analog with low affinity for Ca2+ . Consequently, our findings uncover PFKFB3 inhibition as an Ca2+ -independent mechanism through which BAPTAi impairs cellular metabolism and ultimately compromises the survival of MCL-1-dependent cancer cells. These findings hold two important implications. Firstly, the direct inhibition of PFKFB3 emerges as a key regulator of mTORC1 activity and a promising target in MCL-1-dependent cancers. Secondly, cellular effects caused by BAPTAi are not necessarily related to Ca2+ signaling. Our data support the need for a reassessment of the role of Ca2+ in cellular processes when findings were based on the use of BAPTAi ., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
26. VEGF-B prevents excessive angiogenesis by inhibiting FGF2/FGFR1 pathway.
- Author
-
Lee C, Chen R, Sun G, Liu X, Lin X, He C, Xing L, Liu L, Jensen LD, Kumar A, Langer HF, Ren X, Zhang J, Huang L, Yin X, Kim J, Zhu J, Huang G, Li J, Lu W, Chen W, Liu J, Hu J, Sun Q, Lu W, Fang L, Wang S, Kuang H, Zhang Y, Tian G, Mi J, Kang BA, Narazaki M, Prodeus A, Schoonjans L, Ornitz DM, Gariepy J, Eelen G, Dewerchin M, Yang Y, Ou JS, Mora A, Yao J, Zhao C, Liu Y, Carmeliet P, Cao Y, and Li X
- Subjects
- Humans, Immunotherapy, Receptor, Fibroblast Growth Factor, Type 1 genetics, Fibroblast Growth Factor 2 genetics, Vascular Endothelial Growth Factor B
- Abstract
Although VEGF-B was discovered as a VEGF-A homolog a long time ago, the angiogenic effect of VEGF-B remains poorly understood with limited and diverse findings from different groups. Notwithstanding, drugs that inhibit VEGF-B together with other VEGF family members are being used to treat patients with various neovascular diseases. It is therefore critical to have a better understanding of the angiogenic effect of VEGF-B and the underlying mechanisms. Using comprehensive in vitro and in vivo methods and models, we reveal here for the first time an unexpected and surprising function of VEGF-B as an endogenous inhibitor of angiogenesis by inhibiting the FGF2/FGFR1 pathway when the latter is abundantly expressed. Mechanistically, we unveil that VEGF-B binds to FGFR1, induces FGFR1/VEGFR1 complex formation, and suppresses FGF2-induced Erk activation, and inhibits FGF2-driven angiogenesis and tumor growth. Our work uncovers a previously unrecognized novel function of VEGF-B in tethering the FGF2/FGFR1 pathway. Given the anti-angiogenic nature of VEGF-B under conditions of high FGF2/FGFR1 levels, caution is warranted when modulating VEGF-B activity to treat neovascular diseases., (© 2023. West China Hospital, Sichuan University.)
- Published
- 2023
- Full Text
- View/download PDF
27. Prioritization and functional validation of target genes from single-cell transcriptomics studies.
- Author
-
Sokol L, Cuypers A, Truong AK, Bouché A, Brepoels K, Souffreau J, Rohlenova K, Vinckier S, Schoonjans L, Eelen G, Dewerchin M, de Rooij LPMH, and Carmeliet P
- Subjects
- Transcriptome, Gene Expression Profiling methods
- Abstract
Translation of academic results into clinical practice is a formidable unmet medical need. Single-cell RNA-sequencing (scRNA-seq) studies generate long descriptive ranks of markers with predicted biological function, but without functional validation, it remains challenging to know which markers truly exert the putative function. Given the lengthy/costly nature of validation studies, gene prioritization is required to select candidates. We address these issues by studying tip endothelial cell (EC) marker genes because of their importance for angiogenesis. Here, by tailoring Guidelines On Target Assessment for Innovative Therapeutics, we in silico prioritize previously unreported/poorly described, high-ranking tip EC markers. Notably, functional validation reveals that four of six candidates behave as tip EC genes. We even discover a tip EC function for a gene lacking in-depth functional annotation. Thus, validating prioritized genes from scRNA-seq studies offers opportunities for identifying targets to be considered for possible translation, but not all top-ranked scRNA-seq markers exert the predicted function., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
28. The pulmonary vasculature in lethal COVID-19 and idiopathic pulmonary fibrosis at single-cell resolution.
- Author
-
de Rooij LPMH, Becker LM, Teuwen LA, Boeckx B, Jansen S, Feys S, Verleden S, Liesenborghs L, Stalder AK, Libbrecht S, Van Buyten T, Philips G, Subramanian A, Dumas SJ, Meta E, Borri M, Sokol L, Dendooven A, Truong AK, Gunst J, Van Mol P, Haslbauer JD, Rohlenova K, Menter T, Boudewijns R, Geldhof V, Vinckier S, Amersfoort J, Wuyts W, Van Raemdonck D, Jacobs W, Ceulemans LJ, Weynand B, Thienpont B, Lammens M, Kuehnel M, Eelen G, Dewerchin M, Schoonjans L, Jonigk D, van Dorpe J, Tzankov A, Wauters E, Mazzone M, Neyts J, Wauters J, Lambrechts D, and Carmeliet P
- Subjects
- Humans, Lung metabolism, Transcriptome, COVID-19, Idiopathic Pulmonary Fibrosis genetics, Idiopathic Pulmonary Fibrosis metabolism, Respiratory Distress Syndrome metabolism
- Abstract
Aims: Severe acute respiratory syndrome coronavirus-2 infection causes COVID-19, which in severe cases evokes life-threatening acute respiratory distress syndrome (ARDS). Transcriptome signatures and the functional relevance of non-vascular cell types (e.g. immune and epithelial cells) in COVID-19 are becoming increasingly evident. However, despite its known contribution to vascular inflammation, recruitment/invasion of immune cells, vascular leakage, and perturbed haemostasis in the lungs of severe COVID-19 patients, an in-depth interrogation of the endothelial cell (EC) compartment in lethal COVID-19 is lacking. Moreover, progressive fibrotic lung disease represents one of the complications of COVID-19 pneumonia and ARDS. Analogous features between idiopathic pulmonary fibrosis (IPF) and COVID-19 suggest partial similarities in their pathophysiology, yet, a head-to-head comparison of pulmonary cell transcriptomes between both conditions has not been implemented to date., Methods and Results: We performed single-nucleus RNA-sequencing on frozen lungs from 7 deceased COVID-19 patients, 6 IPF explant lungs, and 12 controls. The vascular fraction, comprising 38 794 nuclei, could be subclustered into 14 distinct EC subtypes. Non-vascular cell types, comprising 137 746 nuclei, were subclustered and used for EC-interactome analyses. Pulmonary ECs of deceased COVID-19 patients showed an enrichment of genes involved in cellular stress, as well as signatures suggestive of dampened immunomodulation and impaired vessel wall integrity. In addition, increased abundance of a population of systemic capillary and venous ECs was identified in COVID-19 and IPF. COVID-19 systemic ECs closely resembled their IPF counterparts, and a set of 30 genes was found congruently enriched in systemic ECs across studies. Receptor-ligand interaction analysis of ECs with non-vascular cell types in the pulmonary micro-environment revealed numerous previously unknown interactions specifically enriched/depleted in COVID-19 and/or IPF., Conclusions: This study uncovered novel insights into the abundance, expression patterns, and interactomes of EC subtypes in COVID-19 and IPF, relevant for future investigations into the progression and treatment of both lethal conditions., Competing Interests: Conflict of interest: A.D. received payments from FMC Belgium, and has a leadership/fiduciary role in the Belgian Society of Pathology (non-profit), and European Society of Pathology Nephropathology working group (non-profit). B.T. has a consulting role for ONO pharmaceutical and owns 10X genomics stocks. B.W. received payments from Hologic. L.J.C. received a research grant and consulting fees from MEDTRONIC. S.F. received support from Pfizer for congress attendance. W.W. received research grants and payment for lectures from Roche and Boehringer Ingelheim and a research grant from Galapagos. S.V. received consulting fees from Therakos and Boehringer Ingelheim. J.W. received investigator-initiated grants, consulting fees, speaker fees, and travel grants from Pfizer and Gilead., (© The Author(s) 2022. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2023
- Full Text
- View/download PDF
29. A high-throughput screening campaign against PFKFB3 identified potential inhibitors with novel scaffolds.
- Author
-
Li J, Zhou Y, Eelen G, Zhou QT, Feng WB, Labroska V, Ma FF, Lu HP, Dewerchin M, Carmeliet P, Wang MW, and Yang DH
- Subjects
- Humans, Glycolysis, Human Umbilical Vein Endothelial Cells metabolism, Neovascularization, Pathologic, High-Throughput Screening Assays, Neoplasms metabolism, Phosphofructokinase-2 antagonists & inhibitors, Phosphofructokinase-2 metabolism
- Abstract
The growth of solid tumors depends on tumor vascularization and the endothelial cells (ECs) that line the lumen of blood vessels. ECs generate a large fraction of ATP through glycolysis, and elevation of their glycolytic activity is associated with angiogenic behavior in solid tumors. 6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) positively regulates glycolysis via fructose-2/6-bisphosphate, the product of its kinase activity. Partial inhibition of glycolysis in tumor ECs by targeting PFKFB3 normalizes the otherwise abnormal tumor vessels, thereby reducing metastasis and improving the outcome of chemotherapy. Although a limited number of tool compounds exist, orally available PFKFB3 inhibitors are unavailable. In this study we conducted a high-throughput screening campaign against the kinase activity of PFKFB3, involving 250,240 chemical compounds. A total of 507 initial hits showing >50% inhibition at 20 µM were identified, 66 of them plus 1 analog from a similarity search consistently displayed low IC
50 values (<10 µM). In vitro experiments yielded 22 nontoxic hits that suppressed the tube formation of primary human umbilical vein ECs at 10 µM. Of them, 15 exhibited binding affinity to PFKFB3 in surface plasmon resonance assays, including 3 (WNN0403-E003, WNN1352-H007 and WNN1542-F004) that passed the pan-assay interference compounds screening without warning flags. This study provides potential leads to the development of new PFKFB3 inhibitors., (© 2022. The Author(s), under exclusive licence to Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Chinese Pharmacological Society.)- Published
- 2023
- Full Text
- View/download PDF
30. Generation of vessel co-option lung metastases mouse models for single-cell isolation of metastases-derived cells and endothelial cells.
- Author
-
Cuypers A, Teuwen LA, Bridgeman VL, de Rooij LPMH, Eelen G, Dewerchin M, Cantelmo AR, Kalucka J, Bouché A, Vinckier S, Carton A, Manderveld A, Vermeulen PB, Reynolds AR, and Carmeliet P
- Subjects
- Mice, Animals, Endothelial Cells, Disease Models, Animal, Neovascularization, Pathologic pathology, Lung Neoplasms pathology
- Abstract
Tumor vessel co-option, a process in which cancer cells "hijack" pre-existing blood vessels to grow and invade healthy tissue, is poorly understood but is a proposed resistance mechanism against anti-angiogenic therapy (AAT). Here, we describe protocols for establishing murine renal (RENCA) and breast (4T1) cancer lung vessel co-option metastases models. Moreover, we outline a reproducible protocol for single-cell isolation from murine lung metastases using magnetic-activated cell sorting as well as immunohistochemical stainings to distinguish vessel co-option from angiogenesis. For complete details on the use and execution of this protocol, please refer to Teuwen et al. (2021)., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
31. Mitochondrial respiration supports autophagy to provide stress resistance during quiescence.
- Author
-
Magalhaes-Novais S, Blecha J, Naraine R, Mikesova J, Abaffy P, Pecinova A, Milosevic M, Bohuslavova R, Prochazka J, Khan S, Novotna E, Sindelka R, Machan R, Dewerchin M, Vlcak E, Kalucka J, Stemberkova Hubackova S, Benda A, Goveia J, Mracek T, Barinka C, Carmeliet P, Neuzil J, Rohlenova K, and Rohlena J
- Subjects
- AMP-Activated Protein Kinases metabolism, Adenosine Triphosphate metabolism, Animals, Cysteine metabolism, DNA, Mitochondrial metabolism, Dextrans metabolism, Endothelial Cells metabolism, Fibroblasts metabolism, Formaldehyde metabolism, Humans, Isothiocyanates, Lipopolysaccharides metabolism, Mechanistic Target of Rapamycin Complex 1 metabolism, Mice, Microtubule-Associated Proteins metabolism, Mitochondria metabolism, Phosphatidylethanolamines metabolism, Reactive Oxygen Species metabolism, Respiration, Sirolimus, Autophagy, Inflammatory Bowel Diseases metabolism
- Abstract
Mitochondrial oxidative phosphorylation (OXPHOS) generates ATP, but OXPHOS also supports biosynthesis during proliferation. In contrast, the role of OXPHOS during quiescence, beyond ATP production, is not well understood. Using mouse models of inducible OXPHOS deficiency in all cell types or specifically in the vascular endothelium that negligibly relies on OXPHOS-derived ATP, we show that selectively during quiescence OXPHOS provides oxidative stress resistance by supporting macroautophagy/autophagy. Mechanistically, OXPHOS constitutively generates low levels of endogenous ROS that induce autophagy via attenuation of ATG4B activity, which provides protection from ROS insult. Physiologically, the OXPHOS-autophagy system (i) protects healthy tissue from toxicity of ROS-based anticancer therapy, and (ii) provides ROS resistance in the endothelium, ameliorating systemic LPS-induced inflammation as well as inflammatory bowel disease. Hence, cells acquired mitochondria during evolution to profit from oxidative metabolism, but also built in an autophagy-based ROS-induced protective mechanism to guard against oxidative stress associated with OXPHOS function during quiescence. Abbreviations : AMPK: AMP-activated protein kinase; AOX: alternative oxidase; Baf A: bafilomycin A
1 ; CI, respiratory complexes I; DCF-DA: 2',7'-dichlordihydrofluorescein diacetate; DHE: dihydroethidium; DSS: dextran sodium sulfate; ΔΨmi: mitochondrial inner membrane potential; EdU: 5-ethynyl-2'-deoxyuridine; ETC: electron transport chain; FA: formaldehyde; HUVEC; human umbilical cord endothelial cells; IBD: inflammatory bowel disease; LC3B: microtubule associated protein 1 light chain 3 beta; LPS: lipopolysaccharide; MEFs: mouse embryonic fibroblasts; MTORC1: mechanistic target of rapamycin kinase complex 1; mtDNA: mitochondrial DNA; NAC: N-acetyl cysteine; OXPHOS: oxidative phosphorylation; PCs: proliferating cells; PE: phosphatidylethanolamine; PEITC: phenethyl isothiocyanate; QCs: quiescent cells; ROS: reactive oxygen species; PLA2: phospholipase A2, WB: western blot.- Published
- 2022
- Full Text
- View/download PDF
32. Single cell atlas identifies lipid-processing and immunomodulatory endothelial cells in healthy and malignant breast.
- Author
-
Geldhof V, de Rooij LPMH, Sokol L, Amersfoort J, De Schepper M, Rohlenova K, Hoste G, Vanderstichele A, Delsupehe AM, Isnaldi E, Dai N, Taverna F, Khan S, Truong AK, Teuwen LA, Richard F, Treps L, Smeets A, Nevelsteen I, Weynand B, Vinckier S, Schoonjans L, Kalucka J, Desmedt C, Neven P, Mazzone M, Floris G, Punie K, Dewerchin M, Eelen G, Wildiers H, Li X, Luo Y, and Carmeliet P
- Subjects
- Endothelial Cells pathology, Female, Humans, Immunity, Ligands, Lipids, PPAR gamma genetics, RNA, Retrospective Studies, Breast Neoplasms pathology, Metformin pharmacology
- Abstract
Since a detailed inventory of endothelial cell (EC) heterogeneity in breast cancer (BC) is lacking, here we perform single cell RNA-sequencing of 26,515 cells (including 8433 ECs) from 9 BC patients and compare them to published EC taxonomies from lung tumors. Angiogenic ECs are phenotypically similar, while other EC subtypes are different. Predictive interactome analysis reveals known but also previously unreported receptor-ligand interactions between ECs and immune cells, suggesting an involvement of breast EC subtypes in immune responses. We also identify a capillary EC subtype (LIPEC (Lipid Processing EC)), which expresses genes involved in lipid processing that are regulated by PPAR-γ and is more abundant in peri-tumoral breast tissue. Retrospective analysis of 4648 BC patients reveals that treatment with metformin (an indirect PPAR-γ signaling activator) provides long-lasting clinical benefit and is positively associated with LIPEC abundance. Our findings warrant further exploration of this LIPEC/PPAR-γ link for BC treatment., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
33. Lipid droplet degradation by autophagy connects mitochondria metabolism to Prox1-driven expression of lymphatic genes and lymphangiogenesis.
- Author
-
Meçe O, Houbaert D, Sassano ML, Durré T, Maes H, Schaaf M, More S, Ganne M, García-Caballero M, Borri M, Verhoeven J, Agrawal M, Jacobs K, Bergers G, Blacher S, Ghesquière B, Dewerchin M, Swinnen JV, Vinckier S, Soengas MS, Carmeliet P, Noël A, and Agostinis P
- Subjects
- Animals, Autophagy genetics, Endothelial Cells metabolism, Fatty Acids metabolism, Lipid Droplets metabolism, Mice, Mitochondria, Transcription Factors metabolism, Lymphangiogenesis genetics, Lymphatic Vessels metabolism
- Abstract
Autophagy has vasculoprotective roles, but whether and how it regulates lymphatic endothelial cells (LEC) homeostasis and lymphangiogenesis is unknown. Here, we show that genetic deficiency of autophagy in LEC impairs responses to VEGF-C and injury-driven corneal lymphangiogenesis. Autophagy loss in LEC compromises the expression of main effectors of LEC identity, like VEGFR3, affects mitochondrial dynamics and causes an accumulation of lipid droplets (LDs) in vitro and in vivo. When lipophagy is impaired, mitochondrial ATP production, fatty acid oxidation, acetyl-CoA/CoA ratio and expression of lymphangiogenic PROX1 target genes are dwindled. Enforcing mitochondria fusion by silencing dynamin-related-protein 1 (DRP1) in autophagy-deficient LEC fails to restore LDs turnover and lymphatic gene expression, whereas supplementing the fatty acid precursor acetate rescues VEGFR3 levels and signaling, and lymphangiogenesis in LEC-Atg5
-/- mice. Our findings reveal that lipophagy in LEC by supporting FAO, preserves a mitochondrial-PROX1 gene expression circuit that safeguards LEC responsiveness to lymphangiogenic mediators and lymphangiogenesis., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
34. Protocols for endothelial cell isolation from mouse tissues: brain, choroid, lung, and muscle.
- Author
-
Conchinha NV, Sokol L, Teuwen LA, Veys K, Dumas SJ, Meta E, García-Caballero M, Geldhof V, Chen R, Treps L, Borri M, de Zeeuw P, Falkenberg KD, Dubois C, Parys M, de Rooij LPMH, Rohlenova K, Goveia J, Schoonjans L, Dewerchin M, Eelen G, Li X, Kalucka J, and Carmeliet P
- Subjects
- Animals, Flow Cytometry methods, Male, Mice, Mice, Inbred C57BL, Brain cytology, Choroid cytology, Endothelial Cells cytology, Lung cytology, Muscles cytology
- Abstract
Endothelial cells (ECs) harbor distinct phenotypical and functional characteristics depending on their tissue localization and contribute to brain, eye, lung, and muscle diseases such as dementia, macular degeneration, pulmonary hypertension, and sarcopenia. To study their function, isolation of pure ECs in high quantities is crucial. Here, we describe protocols for rapid and reproducible blood vessel EC purification established for scRNA sequencing from murine tissues using mechanical and enzymatic digestion followed by magnetic and fluorescence-activated cell sorting. For complete details on the use and execution of these protocol, please refer to Kalucka et al. (2020), Rohlenova et al. (2020), and Goveia et al. (2020)., Competing Interests: The authors declare no competing interests., (© 2021 The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
35. Combined glucocorticoid resistance and hyperlactatemia contributes to lethal shock in sepsis.
- Author
-
Vandewalle J, Timmermans S, Paakinaho V, Vancraeynest L, Dewyse L, Vanderhaeghen T, Wallaeys C, Van Wyngene L, Van Looveren K, Nuyttens L, Eggermont M, Dewaele S, Velho TR, Moita LF, Weis S, Sponholz C, van Grunsven LA, Dewerchin M, Carmeliet P, De Bosscher K, Van de Voorde J, Palvimo JJ, and Libert C
- Subjects
- Animals, Glucocorticoids, Lactic Acid, Mice, Receptors, Glucocorticoid metabolism, Vascular Endothelial Growth Factor A, Hyperlactatemia, Sepsis complications, Sepsis metabolism
- Abstract
Sepsis is a potentially lethal syndrome resulting from a maladaptive response to infection. Upon infection, glucocorticoids are produced as a part of the compensatory response to tolerate sepsis. This tolerance is, however, mitigated in sepsis due to a quickly induced glucocorticoid resistance at the level of the glucocorticoid receptor. Here, we show that defects in the glucocorticoid receptor signaling pathway aggravate sepsis pathophysiology by lowering lactate clearance and sensitizing mice to lactate-induced toxicity. The latter is exerted via an uncontrolled production of vascular endothelial growth factor, resulting in vascular leakage and collapse with severe hypotension, organ damage, and death, all being typical features of a lethal form of sepsis. In conclusion, sepsis leads to glucocorticoid receptor failure and hyperlactatemia, which collectively leads to a lethal vascular collapse., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
36. Tissue factor cytoplasmic domain exacerbates post-infarct left ventricular remodeling via orchestrating cardiac inflammation and angiogenesis.
- Author
-
Chong SY, Zharkova O, Yatim SMJM, Wang X, Lim XC, Huang C, Tan CY, Jiang J, Ye L, Tan MS, Angeli V, Versteeg HH, Dewerchin M, Carmeliet P, Lam CSP, Chan MY, de Kleijn DPV, and Wang JW
- Subjects
- Animals, Cell Proliferation physiology, Inflammation metabolism, Macrophage Activation physiology, Macrophages metabolism, Male, Mice, Mice, Inbred C57BL, Myocardial Infarction metabolism, Myocardium pathology, Myocytes, Cardiac metabolism, Myofibroblasts metabolism, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic prevention & control, Protein Domains physiology, Receptor, PAR-1 metabolism, Receptor, PAR-2 metabolism, Signal Transduction physiology, Thromboplastin physiology, Ventricular Function, Left physiology, Myocardial Infarction pathology, Thromboplastin metabolism, Ventricular Remodeling physiology
- Abstract
The coagulation protein tissue factor (TF) regulates inflammation and angiogenesis via its cytoplasmic domain in infection, cancer and diabetes. While TF is highly abundant in the heart and is implicated in cardiac pathology, the contribution of its cytoplasmic domain to post-infarct myocardial injury and adverse left ventricular (LV) remodeling remains unknown. Methods: Myocardial infarction was induced in wild-type mice or mice lacking the TF cytoplasmic domain (TF∆CT) by occlusion of the left anterior descending coronary artery. Heart function was monitored with echocardiography. Heart tissue was collected at different time-points for histological, molecular and flow cytometry analysis. Results: Compared with wild-type mice, TF∆CT had a higher survival rate during a 28-day follow-up after myocardial infarction. Among surviving mice, TF∆CT mice had better cardiac function and less LV remodeling than wild-type mice. The overall improvement of post-infarct cardiac performance in TF∆CT mice, as revealed by speckle-tracking strain analysis, was attributed to reduced myocardial deformation in the peri-infarct region. Histological analysis demonstrated that TF∆CT hearts had in the infarct area greater proliferation of myofibroblasts and better scar formation. Compared with wild-type hearts, infarcted TF∆CT hearts showed less infiltration of proinflammatory cells with concomitant lower expression of protease-activated receptor-1 (PAR1) - Rac1 axis. In particular, infarcted TF∆CT hearts displayed markedly lower ratios of inflammatory M1 macrophages and reparative M2 macrophages (M1/M2). In vitro experiment with primary macrophages demonstrated that deletion of the TF cytoplasmic domain inhibited macrophage polarization toward the M1 phenotype. Furthermore, infarcted TF∆CT hearts presented markedly higher peri-infarct vessel density associated with enhanced endothelial cell proliferation and higher expression of PAR2 and PAR2-associated pro-angiogenic pathway factors. Finally, the overall cardioprotective effects observed in TF∆CT mice could be abolished by subcutaneously infusing a cocktail of PAR1-activating peptide and PAR2-inhibiting peptide via osmotic minipumps. Conclusions: Our findings demonstrate that the TF cytoplasmic domain exacerbates post-infarct cardiac injury and adverse LV remodeling via differential regulation of inflammation and angiogenesis. Targeted inhibition of the TF cytoplasmic domain-mediated intracellular signaling may ameliorate post-infarct LV remodeling without perturbing coagulation., Competing Interests: Competing Interests: The authors have declared that no competing interest exists., (© The author(s).)
- Published
- 2021
- Full Text
- View/download PDF
37. Protocols for endothelial cell isolation from mouse tissues: kidney, spleen, and testis.
- Author
-
Dumas SJ, Meta E, Conchinha NV, Sokol L, Chen R, Borri M, Teuwen LA, Veys K, García-Caballero M, Geldhof V, Treps L, de Zeeuw P, Falkenberg KD, Dubois C, Parys M, de Rooij LPMH, Rohlenova K, Goveia J, Schoonjans L, Dewerchin M, Eelen G, Li X, Kalucka J, and Carmeliet P
- Subjects
- Animals, Flow Cytometry, Male, Mice, Endothelial Cells cytology, Kidney cytology, Spleen cytology, Testis cytology
- Abstract
Endothelial cells (ECs) exhibit phenotypic and functional tissue specificities, critical for studies in the vascular field and beyond. Thus, tissue-specific methods for isolation of highly purified ECs are necessary. Kidney, spleen, and testis ECs are relevant players in health and diseases such as chronic kidney disease, acute kidney injury, myelofibrosis, and cancer. Here, we provide tailored protocols for rapid and reproducible EC purification established for scRNA sequencing from these adult murine tissues using the combination of magnetic- and fluorescence-activated cell sorting. For complete details on the use and execution of these protocols, please refer to Kalucka et al. (2020) and Dumas et al. (2020)., Competing Interests: The authors declare no competing interests., (© 2021 The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
38. Tumor vessel co-option probed by single-cell analysis.
- Author
-
Teuwen LA, De Rooij LPMH, Cuypers A, Rohlenova K, Dumas SJ, García-Caballero M, Meta E, Amersfoort J, Taverna F, Becker LM, Veiga N, Cantelmo AR, Geldhof V, Conchinha NV, Kalucka J, Treps L, Conradi LC, Khan S, Karakach TK, Soenen S, Vinckier S, Schoonjans L, Eelen G, Van Laere S, Dewerchin M, Dirix L, Mazzone M, Luo Y, Vermeulen P, and Carmeliet P
- Subjects
- Animals, Cell Line, Tumor, Endothelial Cells pathology, Female, Kidney Neoplasms pathology, Lung Neoplasms secondary, Macrophages pathology, Mice, Inbred BALB C, Myeloid Cells pathology, Pericytes pathology, Mice, Neoplasms blood supply, Neoplasms pathology, Single-Cell Analysis
- Abstract
Tumor vessel co-option is poorly understood, yet it is a resistance mechanism against anti-angiogenic therapy (AAT). The heterogeneity of co-opted endothelial cells (ECs) and pericytes, co-opting cancer and myeloid cells in tumors growing via vessel co-option, has not been investigated at the single-cell level. Here, we use a murine AAT-resistant lung tumor model, in which VEGF-targeting induces vessel co-option for continued growth. Single-cell RNA sequencing (scRNA-seq) of 31,964 cells reveals, unexpectedly, a largely similar transcriptome of co-opted tumor ECs (TECs) and pericytes as their healthy counterparts. Notably, we identify cell types that might contribute to vessel co-option, i.e., an invasive cancer-cell subtype, possibly assisted by a matrix-remodeling macrophage population, and another M1-like macrophage subtype, possibly involved in keeping or rendering vascular cells quiescent., Competing Interests: Declaration of interest The authors declare no competing interests., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
39. Protocols for endothelial cell isolation from mouse tissues: small intestine, colon, heart, and liver.
- Author
-
Sokol L, Geldhof V, García-Caballero M, Conchinha NV, Dumas SJ, Meta E, Teuwen LA, Veys K, Chen R, Treps L, Borri M, de Zeeuw P, Falkenberg KD, Dubois C, Parys M, de Rooij LPMH, Goveia J, Rohlenova K, Schoonjans L, Dewerchin M, Eelen G, Li X, Kalucka J, and Carmeliet P
- Subjects
- Animals, Cells, Cultured, Male, Mice, Mice, Inbred C57BL, Endothelial Cells cytology, Flow Cytometry methods, Intestines cytology, Liver cytology, Myocardium cytology
- Abstract
Endothelial cells (ECs) from the small intestine, colon, liver, and heart have distinct phenotypes and functional adaptations that are dependent on their physiological environment. Gut ECs adapt to low oxygen, heart ECs to contractile forces, and liver ECs to low flow rates. Isolating high-purity ECs in sufficient quantities is crucial to study their functions. Here, we describe protocols combining magnetic and fluorescent activated cell sorting for rapid and reproducible EC purification from four adult murine tissues. For complete details on the use and execution of these protocols, please refer to Kalucka et al. (2020)., Competing Interests: The authors declare no competing interests., (© 2021 The Authors.)
- Published
- 2021
- Full Text
- View/download PDF
40. Transcriptomic analysis of CFTR-impaired endothelial cells reveals a pro-inflammatory phenotype.
- Author
-
Declercq M, de Zeeuw P, Conchinha NV, Geldhof V, Ramalho AS, García-Caballero M, Brepoels K, Ensinck M, Carlon MS, Bird MJ, Vinckier S, Proesmans M, Vermeulen F, Dupont L, Ghesquière B, Dewerchin M, Carmeliet P, Cassiman D, Treps L, Eelen G, and Witters P
- Subjects
- Endothelial Cells metabolism, Humans, Phenotype, Transcriptome, Cystic Fibrosis genetics, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Cystic Fibrosis Transmembrane Conductance Regulator metabolism
- Abstract
Cystic fibrosis (CF) is a life-threatening disorder characterised by decreased pulmonary mucociliary and pathogen clearance, and an exaggerated inflammatory response leading to progressive lung damage. CF is caused by bi-allelic pathogenic variants of the cystic fibrosis transmembrane conductance regulator (CFTR) gene, which encodes a chloride channel. CFTR is expressed in endothelial cells (ECs) and EC dysfunction has been reported in CF patients, but a role for this ion channel in ECs regarding CF disease progression is poorly described.We used an unbiased RNA sequencing approach in complementary models of CFTR silencing and blockade (by the CFTR inhibitor CFTRinh-172) in human ECs to characterise the changes upon CFTR impairment. Key findings were further validated in vitro and in vivo in CFTR-knockout mice and ex vivo in CF patient-derived ECs.Both models of CFTR impairment revealed that EC proliferation, migration and autophagy were downregulated. Remarkably though, defective CFTR function led to EC activation and a persisting pro-inflammatory state of the endothelium with increased leukocyte adhesion. Further validation in CFTR-knockout mice revealed enhanced leukocyte extravasation in lung and liver parenchyma associated with increased levels of EC activation markers. In addition, CF patient-derived ECs displayed increased EC activation markers and leukocyte adhesion, which was partially rescued by the CFTR modulators VX-770 and VX-809.Our integrated analysis thus suggests that ECs are no innocent bystanders in CF pathology, but rather may contribute to the exaggerated inflammatory phenotype, raising the question of whether normalisation of vascular inflammation might be a novel therapeutic strategy to ameliorate the disease severity of CF., Competing Interests: Conflict of interest: M. Declercq has nothing to disclose. Conflict of interest: P. de Zeeuw has nothing to disclose. Conflict of interest: N.V. Conchinha has nothing to disclose. Conflict of interest: V. Geldhof has nothing to disclose. Conflict of interest: A.S. Ramalho has nothing to disclose. Conflict of interest: M. García-Caballero has nothing to disclose. Conflict of interest: K. Brepoels has nothing to disclose. Conflict of interest: M. Ensinck has nothing to disclose. Conflict of interest: M.S. Carlon has nothing to disclose. Conflict of interest: M.J. Bird has nothing to disclose. Conflict of interest: S. Vinckier has nothing to disclose. Conflict of interest: M. Proesmans has nothing to disclose. Conflict of interest: F. Vermeulen has nothing to disclose. Conflict of interest: L. Dupont has nothing to disclose. Conflict of interest: B. Ghesquière has nothing to disclose. Conflict of interest: M. Dewerchin has nothing to disclose. Conflict of interest: P. Carmeliet has nothing to disclose. Conflict of interest: D. Cassiman has nothing to disclose. Conflict of interest: L. Treps has nothing to disclose. Conflict of interest: G. Eelen has nothing to disclose. Conflict of interest: P. Witters has nothing to disclose., (Copyright ©ERS 2021.)
- Published
- 2021
- Full Text
- View/download PDF
41. BIOMEX: an interactive workflow for (single cell) omics data interpretation and visualization.
- Author
-
Taverna F, Goveia J, Karakach TK, Khan S, Rohlenova K, Treps L, Subramanian A, Schoonjans L, Dewerchin M, Eelen G, and Carmeliet P
- Subjects
- Algorithms, Bile Duct Neoplasms genetics, Cholangiocarcinoma genetics, Computer Graphics, Endothelial Cells metabolism, Humans, Metabolomics methods, Neoplasms mortality, Proteomics methods, Survival Analysis, Workflow, Gene Expression Profiling methods, Single-Cell Analysis methods, Software
- Abstract
The amount of biological data, generated with (single cell) omics technologies, is rapidly increasing, thereby exacerbating bottlenecks in the data analysis and interpretation of omics experiments. Data mining platforms that facilitate non-bioinformatician experimental scientists to analyze a wide range of experimental designs and data types can alleviate such bottlenecks, aiding in the exploration of (newly generated or publicly available) omics datasets. Here, we present BIOMEX, a browser-based software, designed to facilitate the Biological Interpretation Of Multi-omics EXperiments by bench scientists. BIOMEX integrates state-of-the-art statistical tools and field-tested algorithms into a flexible but well-defined workflow that accommodates metabolomics, transcriptomics, proteomics, mass cytometry and single cell data from different platforms and organisms. The BIOMEX workflow is accompanied by a manual and video tutorials that provide the necessary background to navigate the interface and get acquainted with the employed methods. BIOMEX guides the user through omics-tailored analyses, such as data pretreatment and normalization, dimensionality reduction, differential and enrichment analysis, pathway mapping, clustering, marker analysis, trajectory inference, meta-analysis and others. BIOMEX is fully interactive, allowing users to easily change parameters and generate customized plots exportable as high-quality publication-ready figures. BIOMEX is open source and freely available at https://www.vibcancer.be/software-tools/biomex., (© The Author(s) 2020. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2020
- Full Text
- View/download PDF
42. Single-Cell RNA Sequencing Maps Endothelial Metabolic Plasticity in Pathological Angiogenesis.
- Author
-
Rohlenova K, Goveia J, García-Caballero M, Subramanian A, Kalucka J, Treps L, Falkenberg KD, de Rooij LPMH, Zheng Y, Lin L, Sokol L, Teuwen LA, Geldhof V, Taverna F, Pircher A, Conradi LC, Khan S, Stegen S, Panovska D, De Smet F, Staal FJT, Mclaughlin RJ, Vinckier S, Van Bergen T, Ectors N, De Haes P, Wang J, Bolund L, Schoonjans L, Karakach TK, Yang H, Carmeliet G, Liu Y, Thienpont B, Dewerchin M, Eelen G, Li X, Luo Y, and Carmeliet P
- Subjects
- Animals, Endothelial Cells cytology, Endothelial Cells pathology, HEK293 Cells, Human Umbilical Vein Endothelial Cells, Humans, Male, Mice, Mice, Inbred C57BL, Sequence Analysis, RNA, Single-Cell Analysis, Endothelial Cells metabolism, Lung Neoplasms metabolism, Macular Degeneration metabolism, Neovascularization, Pathologic metabolism, Transcriptome
- Abstract
Endothelial cell (EC) metabolism is an emerging target for anti-angiogenic therapy in tumor angiogenesis and choroidal neovascularization (CNV), but little is known about individual EC metabolic transcriptomes. By single-cell RNA sequencing 28,337 murine choroidal ECs (CECs) and sprouting CNV-ECs, we constructed a taxonomy to characterize their heterogeneity. Comparison with murine lung tumor ECs (TECs) revealed congruent marker gene expression by distinct EC phenotypes across tissues and diseases, suggesting similar angiogenic mechanisms. Trajectory inference predicted that differentiation of venous to angiogenic ECs was accompanied by metabolic transcriptome plasticity. ECs displayed metabolic transcriptome heterogeneity during cell-cycle progression and in quiescence. Hypothesizing that conserved genes are important, we used an integrated analysis, based on congruent transcriptome analysis, CEC-tailored genome-scale metabolic modeling, and gene expression meta-analysis in cross-species datasets, followed by in vitro and in vivo validation, to identify SQLE and ALDH18A1 as previously unknown metabolic angiogenic targets., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
43. An Integrated Gene Expression Landscape Profiling Approach to Identify Lung Tumor Endothelial Cell Heterogeneity and Angiogenic Candidates.
- Author
-
Goveia J, Rohlenova K, Taverna F, Treps L, Conradi LC, Pircher A, Geldhof V, de Rooij LPMH, Kalucka J, Sokol L, García-Caballero M, Zheng Y, Qian J, Teuwen LA, Khan S, Boeckx B, Wauters E, Decaluwé H, De Leyn P, Vansteenkiste J, Weynand B, Sagaert X, Verbeken E, Wolthuis A, Topal B, Everaerts W, Bohnenberger H, Emmert A, Panovska D, De Smet F, Staal FJT, Mclaughlin RJ, Impens F, Lagani V, Vinckier S, Mazzone M, Schoonjans L, Dewerchin M, Eelen G, Karakach TK, Yang H, Wang J, Bolund L, Lin L, Thienpont B, Li X, Lambrechts D, Luo Y, and Carmeliet P
- Published
- 2020
- Full Text
- View/download PDF
44. Towards a widespread adoption of metabolic modeling tools in biopharmaceutical industry: a process systems biology engineering perspective.
- Author
-
Richelle A, David B, Demaegd D, Dewerchin M, Kinet R, Morreale A, Portela R, Zune Q, and von Stosch M
- Subjects
- Biological Products pharmacology, Biotechnology methods, Biotechnology trends, Industry trends, Models, Biological, Bioengineering methods, Biological Products metabolism, Systems Biology methods
- Abstract
In biotechnology, the emergence of high-throughput technologies challenges the interpretation of large datasets. One way to identify meaningful outcomes impacting process and product attributes from large datasets is using systems biology tools such as metabolic models. However, these tools are still not fully exploited for this purpose in industrial context due to gaps in our knowledge and technical limitations. In this paper, key aspects restraining the routine implementation of these tools are highlighted in three research fields: monitoring, network science and hybrid modeling. Advances in these fields could expand the current state of systems biology applications in biopharmaceutical industry to address existing challenges in bioprocess development and improvement.
- Published
- 2020
- Full Text
- View/download PDF
45. Single-Cell Transcriptome Atlas of Murine Endothelial Cells.
- Author
-
Kalucka J, de Rooij LPMH, Goveia J, Rohlenova K, Dumas SJ, Meta E, Conchinha NV, Taverna F, Teuwen LA, Veys K, García-Caballero M, Khan S, Geldhof V, Sokol L, Chen R, Treps L, Borri M, de Zeeuw P, Dubois C, Karakach TK, Falkenberg KD, Parys M, Yin X, Vinckier S, Du Y, Fenton RA, Schoonjans L, Dewerchin M, Eelen G, Thienpont B, Lin L, Bolund L, Li X, Luo Y, and Carmeliet P
- Subjects
- Animals, Brain cytology, Cardiovascular System cytology, Endothelial Cells classification, Endothelial Cells cytology, Gastrointestinal Tract cytology, Male, Mice, Mice, Inbred C57BL, Muscles cytology, Organ Specificity, RNA-Seq, Testis cytology, Endothelial Cells metabolism, Single-Cell Analysis, Transcriptome
- Abstract
The heterogeneity of endothelial cells (ECs) across tissues remains incompletely inventoried. We constructed an atlas of >32,000 single-EC transcriptomes from 11 mouse tissues and identified 78 EC subclusters, including Aqp7
+ intestinal capillaries and angiogenic ECs in healthy tissues. ECs from brain/testis, liver/spleen, small intestine/colon, and skeletal muscle/heart pairwise expressed partially overlapping marker genes. Arterial, venous, and lymphatic ECs shared more markers in more tissues than did heterogeneous capillary ECs. ECs from different vascular beds (arteries, capillaries, veins, lymphatics) exhibited transcriptome similarity across tissues, but the tissue (rather than the vessel) type contributed to the EC heterogeneity. Metabolic transcriptome analysis revealed a similar tissue-grouping phenomenon of ECs and heterogeneous metabolic gene signatures in ECs between tissues and between vascular beds within a single tissue in a tissue-type-dependent pattern. The EC atlas taxonomy enabled identification of EC subclusters in public scRNA-seq datasets and provides a powerful discovery tool and resource value., Competing Interests: Declaration of Interests None of the authors have competing financial interests to declare., (Copyright © 2020 Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
46. Loss or Inhibition of Stromal-Derived PlGF Prolongs Survival of Mice with Imatinib-Resistant Bcr-Abl1 + Leukemia.
- Author
-
Schmidt T, Kharabi Masouleh B, Loges S, Cauwenberghs S, Fraisl P, Maes C, Jonckx B, De Keersmaecker K, Kleppe M, Tjwa M, Schenk T, Vinckier S, Fragoso R, De Mol M, Beel K, Dias S, Verfaillie C, Clark RE, Brümmendorf TH, Vandenberghe P, Rafii S, Holyoake T, Hochhaus A, Cools J, Karin M, Carmeliet G, Dewerchin M, and Carmeliet P
- Published
- 2020
- Full Text
- View/download PDF
47. Single-Cell RNA Sequencing Reveals Renal Endothelium Heterogeneity and Metabolic Adaptation to Water Deprivation.
- Author
-
Dumas SJ, Meta E, Borri M, Goveia J, Rohlenova K, Conchinha NV, Falkenberg K, Teuwen LA, de Rooij L, Kalucka J, Chen R, Khan S, Taverna F, Lu W, Parys M, De Legher C, Vinckier S, Karakach TK, Schoonjans L, Lin L, Bolund L, Dewerchin M, Eelen G, Rabelink TJ, Li X, Luo Y, and Carmeliet P
- Subjects
- Animals, Endothelial Cells physiology, Male, Mice, Mice, Inbred C57BL, Phenotype, Adaptation, Physiological genetics, Endothelial Cells metabolism, Kidney cytology, Sequence Analysis, RNA, Water Deprivation physiology
- Abstract
Background: Renal endothelial cells from glomerular, cortical, and medullary kidney compartments are exposed to different microenvironmental conditions and support specific kidney processes. However, the heterogeneous phenotypes of these cells remain incompletely inventoried. Osmotic homeostasis is vitally important for regulating cell volume and function, and in mammals, osmotic equilibrium is regulated through the countercurrent system in the renal medulla, where water exchange through endothelium occurs against an osmotic pressure gradient. Dehydration exposes medullary renal endothelial cells to extreme hyperosmolarity, and how these cells adapt to and survive in this hypertonic milieu is unknown., Methods: We inventoried renal endothelial cell heterogeneity by single-cell RNA sequencing >40,000 mouse renal endothelial cells, and studied transcriptome changes during osmotic adaptation upon water deprivation. We validated our findings by immunostaining and functionally by targeting oxidative phosphorylation in a hyperosmolarity model in vitro and in dehydrated mice in vivo ., Results: We identified 24 renal endothelial cell phenotypes (of which eight were novel), highlighting extensive heterogeneity of these cells between and within the cortex, glomeruli, and medulla. In response to dehydration and hypertonicity, medullary renal endothelial cells upregulated the expression of genes involved in the hypoxia response, glycolysis, and-surprisingly-oxidative phosphorylation. Endothelial cells increased oxygen consumption when exposed to hyperosmolarity, whereas blocking oxidative phosphorylation compromised endothelial cell viability during hyperosmotic stress and impaired urine concentration during dehydration., Conclusions: This study provides a high-resolution atlas of the renal endothelium and highlights extensive renal endothelial cell phenotypic heterogeneity, as well as a previously unrecognized role of oxidative phosphorylation in the metabolic adaptation of medullary renal endothelial cells to water deprivation., (Copyright © 2020 by the American Society of Nephrology.)
- Published
- 2020
- Full Text
- View/download PDF
48. Role and therapeutic potential of dietary ketone bodies in lymph vessel growth.
- Author
-
García-Caballero M, Zecchin A, Souffreau J, Truong AK, Teuwen LA, Vermaelen W, Martín-Pérez R, de Zeeuw P, Bouché A, Vinckier S, Cornelissen I, Eelen G, Ghesquière B, Mazzone M, Dewerchin M, and Carmeliet P
- Subjects
- Animals, Diet, Ketogenic, Humans, Mice, Oxidation-Reduction, Diet, Ketone Bodies metabolism, Lymphatic Vessels metabolism
- Abstract
Lymphatic vessels (LVs), lined by lymphatic endothelial cells (LECs), are indispensable for life
1 . However, the role of metabolism in LECs has been incompletely elucidated. In the present study, it is reported that LEC-specific loss of OXCT1, a key enzyme of ketone body oxidation2 , reduces LEC proliferation, migration and vessel sprouting in vitro and impairs lymphangiogenesis in development and disease in Prox1ΔOXCT1 mice. Mechanistically, OXCT1 silencing lowers acetyl-CoA levels, tricarboxylic acid cycle metabolite pools, and nucleotide precursor and deoxynucleotide triphosphate levels required for LEC proliferation. Ketone body supplementation to LECs induces the opposite effects. Notably, elevation of lymph ketone body levels by a high-fat, low-carbohydrate ketogenic diet or by administration of the ketone body β-hydroxybutyrate increases lymphangiogenesis after corneal injury and myocardial infarction. Intriguingly, in a mouse model of microsurgical ablation of LVs in the tail, which repeats features of acquired lymphoedema in humans, the ketogenic diet improves LV function and growth, reduces infiltration of anti-lymphangiogenic immune cells and decreases oedema, suggesting a novel dietary therapeutic opportunity.- Published
- 2019
- Full Text
- View/download PDF
49. EndoDB: a database of endothelial cell transcriptomics data.
- Author
-
Khan S, Taverna F, Rohlenova K, Treps L, Geldhof V, de Rooij L, Sokol L, Pircher A, Conradi LC, Kalucka J, Schoonjans L, Eelen G, Dewerchin M, Karakach T, Li X, Goveia J, and Carmeliet P
- Subjects
- Animals, Endothelial Cells metabolism, Gene Expression Regulation genetics, Humans, Principal Component Analysis, Computational Biology, Databases, Genetic, Transcriptome genetics
- Abstract
Endothelial cells (ECs) line blood vessels, regulate homeostatic processes (blood flow, immune cell trafficking), but are also involved in many prevalent diseases. The increasing use of high-throughput technologies such as gene expression microarrays and (single cell) RNA sequencing generated a wealth of data on the molecular basis of EC (dys-)function. Extracting biological insight from these datasets is challenging for scientists who are not proficient in bioinformatics. To facilitate the re-use of publicly available EC transcriptomics data, we developed the endothelial database EndoDB, a web-accessible collection of expert curated, quality assured and pre-analyzed data collected from 360 datasets comprising a total of 4741 bulk and 5847 single cell endothelial transcriptomes from six different organisms. Unlike other added-value databases, EndoDB allows to easily retrieve and explore data of specific studies, determine under which conditions genes and pathways of interest are deregulated and assess reprogramming of metabolism via principal component analysis, differential gene expression analysis, gene set enrichment analysis, heatmaps and metabolic and transcription factor analysis, while single cell data are visualized as gene expression color-coded t-SNE plots. Plots and tables in EndoDB are customizable, downloadable and interactive. EndoDB is freely available at https://vibcancer.be/software-tools/endodb, and will be updated to include new studies.
- Published
- 2019
- Full Text
- View/download PDF
50. Impairment of Angiogenesis by Fatty Acid Synthase Inhibition Involves mTOR Malonylation.
- Author
-
Bruning U, Morales-Rodriguez F, Kalucka J, Goveia J, Taverna F, Queiroz KCS, Dubois C, Cantelmo AR, Chen R, Loroch S, Timmerman E, Caixeta V, Bloch K, Conradi LC, Treps L, Staes A, Gevaert K, Tee A, Dewerchin M, Semenkovich CF, Impens F, Schilling B, Verdin E, Swinnen JV, Meier JL, Kulkarni RA, Sickmann A, Ghesquière B, Schoonjans L, Li X, Mazzone M, and Carmeliet P
- Subjects
- Acetyl-CoA Carboxylase antagonists & inhibitors, Animals, Cell Line, Tumor, Cell Proliferation, Fatty Acid Synthase, Type I antagonists & inhibitors, Fatty Acid Synthase, Type I genetics, Human Umbilical Vein Endothelial Cells cytology, Humans, Mechanistic Target of Rapamycin Complex 1 metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Orlistat therapeutic use, Protein Processing, Post-Translational, Retinal Neovascularization drug therapy, Fatty Acid Synthase, Type I physiology, Human Umbilical Vein Endothelial Cells metabolism, Malonyl Coenzyme A metabolism, Retinal Neovascularization pathology, TOR Serine-Threonine Kinases metabolism
- Abstract
The role of fatty acid synthesis in endothelial cells (ECs) remains incompletely characterized. We report that fatty acid synthase knockdown (FASN
KD ) in ECs impedes vessel sprouting by reducing proliferation. Endothelial loss of FASN impaired angiogenesis in vivo, while FASN blockade reduced pathological ocular neovascularization, at >10-fold lower doses than used for anti-cancer treatment. Impaired angiogenesis was not due to energy stress, redox imbalance, or palmitate depletion. Rather, FASNKD elevated malonyl-CoA levels, causing malonylation (a post-translational modification) of mTOR at lysine 1218 (K1218). mTOR K-1218 malonylation impaired mTOR complex 1 (mTORC1) kinase activity, thereby reducing phosphorylation of downstream targets (p70S6K/4EBP1). Silencing acetyl-CoA carboxylase 1 (an enzyme producing malonyl-CoA) normalized malonyl-CoA levels and reactivated mTOR in FASNKD ECs. Mutagenesis unveiled the importance of mTOR K1218 malonylation for angiogenesis. This study unveils a novel role of FASN in metabolite signaling that contributes to explaining the anti-angiogenic effect of FASN blockade., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.